1
|
Munnik C, Xaba MP, Malindisa ST, Russell BL, Sooklal SA. Drosophila melanogaster: A platform for anticancer drug discovery and personalized therapies. Front Genet 2022; 13:949241. [PMID: 36003330 PMCID: PMC9393232 DOI: 10.3389/fgene.2022.949241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is a complex disease whereby multiple genetic aberrations, epigenetic modifications, metabolic reprogramming, and the microenvironment contribute to the development of a tumor. In the traditional anticancer drug discovery pipeline, drug candidates are usually screened in vitro using two-dimensional or three-dimensional cell culture. However, these methods fail to accurately mimic the human disease state. This has led to the poor success rate of anticancer drugs in the preclinical stages since many drugs are abandoned due to inefficacy or toxicity when transitioned to whole-organism models. The common fruit fly, Drosophila melanogaster, has emerged as a beneficial system for modeling human cancers. Decades of fundamental research have shown the evolutionary conservation of key genes and signaling pathways between flies and humans. Moreover, Drosophila has a lower genetic redundancy in comparison to mammals. These factors, in addition to the advancement of genetic toolkits for manipulating gene expression, allow for the generation of complex Drosophila genotypes and phenotypes. Numerous studies have successfully created Drosophila models for colorectal, lung, thyroid, and brain cancers. These models were utilized in the high-throughput screening of FDA-approved drugs which led to the identification of several compounds capable of reducing proliferation and rescuing phenotypes. More noteworthy, Drosophila has also unlocked the potential for personalized therapies. Drosophila ‘avatars’ presenting the same mutations as a patient are used to screen multiple therapeutic agents targeting multiple pathways to find the most appropriate combination of drugs. The outcomes of these studies have translated to significant responses in patients with adenoid cystic carcinoma and metastatic colorectal cancers. Despite not being widely utilized, the concept of in vivo screening of drugs in Drosophila is making significant contributions to the current drug discovery pipeline. In this review, we discuss the application of Drosophila as a platform in anticancer drug discovery; with special focus on the cancer models that have been generated, drug libraries that have been screened and the status of personalized therapies. In addition, we elaborate on the biological and technical limitations of this system.
Collapse
Affiliation(s)
- Chamoné Munnik
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Malungi P. Xaba
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Sibusiso T. Malindisa
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Bonnie L. Russell
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- Buboo (Pty) Ltd, The Innovation Hub, Pretoria, South Africa
| | - Selisha A. Sooklal
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- *Correspondence: Selisha A. Sooklal,
| |
Collapse
|
2
|
Salim S, Banu A, Alwa A, Gowda SBM, Mohammad F. The gut-microbiota-brain axis in autism: what Drosophila models can offer? J Neurodev Disord 2021; 13:37. [PMID: 34525941 PMCID: PMC8442445 DOI: 10.1186/s11689-021-09378-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/28/2022] Open
Abstract
The idea that alterations in gut-microbiome-brain axis (GUMBA)-mediated communication play a crucial role in human brain disorders like autism remains a topic of intensive research in various labs. Gastrointestinal issues are a common comorbidity in patients with autism spectrum disorder (ASD). Although gut microbiome and microbial metabolites have been implicated in the etiology of ASD, the underlying molecular mechanism remains largely unknown. In this review, we have summarized recent findings in human and animal models highlighting the role of the gut-brain axis in ASD. We have discussed genetic and neurobehavioral characteristics of Drosophila as an animal model to study the role of GUMBA in ASD. The utility of Drosophila fruit flies as an amenable genetic tool, combined with axenic and gnotobiotic approaches, and availability of transgenic flies may reveal mechanistic insight into gut-microbiota-brain interactions and the impact of its alteration on behaviors relevant to neurological disorders like ASD.
Collapse
Affiliation(s)
- Safa Salim
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Ayesha Banu
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Amira Alwa
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Swetha B M Gowda
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Farhan Mohammad
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar.
| |
Collapse
|
3
|
Sarkar S, Feany MB. Precision Medicine on the Fly: Using Drosophila to Decipher Gene-Environment Interactions in Parkinson's Disease. Toxicol Sci 2021; 182:159-167. [PMID: 34076689 DOI: 10.1093/toxsci/kfab060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Big data approaches have profoundly influenced state-of-the-art in many fields of research, with toxicology being no exception. Here, we use Parkinson's disease as a window through which to explore the challenges of a dual explosion of metabolomic data addressing the myriad environmental exposures individuals experience and genetic analyses implicating many different loci as risk factors for disease. We argue that new experimental approaches are needed to convert the growing body of omics data into molecular mechanisms of disease that can be therapeutically targeted in specific patients. We outline one attractive strategy, which capitalizes on the rapid generation time and advanced molecular tools available in the fruit fly, Drosophila, to provide a platform for mechanistic dissection and drug discovery.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Gondal MN, Butt RN, Shah OS, Sultan MU, Mustafa G, Nasir Z, Hussain R, Khawar H, Qazi R, Tariq M, Faisal A, Chaudhary SU. A Personalized Therapeutics Approach Using an In Silico Drosophila Patient Model Reveals Optimal Chemo- and Targeted Therapy Combinations for Colorectal Cancer. Front Oncol 2021; 11:692592. [PMID: 34336681 PMCID: PMC8323493 DOI: 10.3389/fonc.2021.692592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
In silico models of biomolecular regulation in cancer, annotated with patient-specific gene expression data, can aid in the development of novel personalized cancer therapeutic strategies. Drosophila melanogaster is a well-established animal model that is increasingly being employed to evaluate such preclinical personalized cancer therapies. Here, we report five Boolean network models of biomolecular regulation in cells lining the Drosophila midgut epithelium and annotate them with colorectal cancer patient-specific mutation data to develop an in silico Drosophila Patient Model (DPM). We employed cell-type-specific RNA-seq gene expression data from the FlyGut-seq database to annotate and then validate these networks. Next, we developed three literature-based colorectal cancer case studies to evaluate cell fate outcomes from the model. Results obtained from analyses of the proposed DPM help: (i) elucidate cell fate evolution in colorectal tumorigenesis, (ii) validate cytotoxicity of nine FDA-approved CRC drugs, and (iii) devise optimal personalized treatment combinations. The personalized network models helped identify synergistic combinations of paclitaxel-regorafenib, paclitaxel-bortezomib, docetaxel-bortezomib, and paclitaxel-imatinib for treating different colorectal cancer patients. Follow-on therapeutic screening of six colorectal cancer patients from cBioPortal using this drug combination demonstrated a 100% increase in apoptosis and a 100% decrease in proliferation. In conclusion, this work outlines a novel roadmap for decoding colorectal tumorigenesis along with the development of personalized combinatorial therapeutics for preclinical translational studies.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rida Nasir Butt
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Osama Shiraz Shah
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Umer Sultan
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ghulam Mustafa
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Zainab Nasir
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Risham Hussain
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Huma Khawar
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Romena Qazi
- Department of Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Muhammad Tariq
- Epigenetics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
5
|
Abstract
AbstractAn important goal in the fight against cancer is to understand how tumors become invasive and metastatic. A crucial early step in metastasis is thought to be the epithelial mesenchymal transition (EMT), the process in which epithelial cells transition into a more migratory and invasive, mesenchymal state. Since the genetic regulatory networks driving EMT in tumors derive from those used in development, analysis of EMTs in genetic model organisms such as the vinegar fly, Drosophila melanogaster, can provide great insight into cancer. In this review I highlight the many ways in which studies in the fly are shedding light on cancer metastasis. The review covers both normal developmental events in which epithelial cells become migratory, as well as induced events, whereby normal epithelial cells become metastatic due to genetic manipulations. The ability to make such precise genetic perturbations in the context of a normal, in vivo environment, complete with a working innate immune system, is making the fly increasingly important in understanding metastasis.
Collapse
Affiliation(s)
- Michael J. Murray
- School of BioSciences, Faculty of Science, University of Melbourne, Victoria 3010, Melbourne, Australia
| |
Collapse
|
6
|
Millet-Boureima C, He S, Le TBU, Gamberi C. Modeling Neoplastic Growth in Renal Cell Carcinoma and Polycystic Kidney Disease. Int J Mol Sci 2021; 22:3918. [PMID: 33920158 PMCID: PMC8070407 DOI: 10.3390/ijms22083918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) and autosomal dominant polycystic kidney disease (ADPKD) share several characteristics, including neoplastic cell growth, kidney cysts, and limited therapeutics. As well, both exhibit impaired vasculature and compensatory VEGF activation of angiogenesis. The PI3K/AKT/mTOR and Ras/Raf/ERK pathways play important roles in regulating cystic and tumor cell proliferation and growth. Both RCC and ADPKD result in hypoxia, where HIF-α signaling is activated in response to oxygen deprivation. Primary cilia and altered cell metabolism may play a role in disease progression. Non-coding RNAs may regulate RCC carcinogenesis and ADPKD through their varied effects. Drosophila exhibits remarkable conservation of the pathways involved in RCC and ADPKD. Here, we review the progress towards understanding disease mechanisms, partially overlapping cellular and molecular dysfunctions in RCC and ADPKD and reflect on the potential for the agile Drosophila genetic model to accelerate discovery science, address unresolved mechanistic aspects of these diseases, and perform rapid pharmacological screens.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Thi Bich Uyen Le
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore 119228, Singapore
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29528-6054, USA
| |
Collapse
|
7
|
Gowda SBM, Salim S, Mohammad F. Anatomy and Neural Pathways Modulating Distinct Locomotor Behaviors in Drosophila Larva. BIOLOGY 2021; 10:90. [PMID: 33504061 PMCID: PMC7910854 DOI: 10.3390/biology10020090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/07/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022]
Abstract
The control of movements is a fundamental feature shared by all animals. At the most basic level, simple movements are generated by coordinated neural activity and muscle contraction patterns that are controlled by the central nervous system. How behavioral responses to various sensory inputs are processed and integrated by the downstream neural network to produce flexible and adaptive behaviors remains an intense area of investigation in many laboratories. Due to recent advances in experimental techniques, many fundamental neural pathways underlying animal movements have now been elucidated. For example, while the role of motor neurons in locomotion has been studied in great detail, the roles of interneurons in animal movements in both basic and noxious environments have only recently been realized. However, the genetic and transmitter identities of many of these interneurons remains unclear. In this review, we provide an overview of the underlying circuitry and neural pathways required by Drosophila larvae to produce successful movements. By improving our understanding of locomotor circuitry in model systems such as Drosophila, we will have a better understanding of how neural circuits in organisms with different bodies and brains lead to distinct locomotion types at the organism level. The understanding of genetic and physiological components of these movements types also provides directions to understand movements in higher organisms.
Collapse
Affiliation(s)
| | | | - Farhan Mohammad
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha 34110, Qatar; (S.B.M.G.); (S.S.)
| |
Collapse
|
8
|
Hamaratoglu F, Atkins M. Rounding up the Usual Suspects: Assessing Yorkie, AP-1, and Stat Coactivation in Tumorigenesis. Int J Mol Sci 2020; 21:E4580. [PMID: 32605129 PMCID: PMC7370090 DOI: 10.3390/ijms21134580] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Can hyperactivation of a few key signaling effectors be the underlying reason for the majority of epithelial cancers despite different driver mutations? Here, to address this question, we use the Drosophila model, which allows analysis of gene expression from tumors with known initiating mutations. Furthermore, its simplified signaling pathways have numerous well characterized targets we can use as pathway readouts. In Drosophila tumor models, changes in the activities of three pathways, Jun N-terminal Kinase (JNK), Janus Kinase / Signal Transducer and Activator of Transcription (JAK/STAT), and Hippo, mediated by AP-1 factors, Stat92E, and Yorkie, are reported frequently. We hypothesized this may indicate that these three pathways are commonly deregulated in tumors. To assess this, we mined the available transcriptomic data and evaluated the activity levels of eight pathways in various tumor models. Indeed, at least two out of our three suspects contribute to tumor development in all Drosophila cancer models assessed, despite different initiating mutations or tissues of origin. Surprisingly, we found that Notch signaling is also globally activated in all models examined. We propose that these four pathways, JNK, JAK/STAT, Hippo, and Notch, are paid special attention and assayed for systematically in existing and newly developed models.
Collapse
Affiliation(s)
| | - Mardelle Atkins
- Department of Biological Sciences, Sam Houston State University, Huntsville, TX 77341, USA
| |
Collapse
|
9
|
Ung PMU, Sonoshita M, Scopton AP, Dar AC, Cagan RL, Schlessinger A. Integrated computational and Drosophila cancer model platform captures previously unappreciated chemicals perturbing a kinase network. PLoS Comput Biol 2019; 15:e1006878. [PMID: 31026276 PMCID: PMC6506148 DOI: 10.1371/journal.pcbi.1006878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 05/08/2019] [Accepted: 02/18/2019] [Indexed: 12/13/2022] Open
Abstract
Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a ‘hybrid’ molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity. Effective and safe treatment of multigenic diseases often involves drugs that address multiple points along disease networks, i.e., polypharmacology. Polypharmacology is increasingly appreciated as a potentially desirable property of kinase drugs. However, most known drugs that interact with multiple targets have been identified as such by chance and most polypharmacological compounds are not chemically unique, resembling structures of known kinase inhibitors. The fruit fly Drosophila provides an inexpensive, rapid, quantitative, whole animal screening platform that has the potential to complement computational approaches. We present a chemical genetics approach that efficiently combines Drosophila with structural prediction and virtual screening, creating a unique discovery platform. We demonstrate the utility of our approach by developing useful small molecules targeting a kinase network in a Drosophila model of Medullary Thyroid Cancer (MTC) driven by oncogenic dRetM955T.
Collapse
Affiliation(s)
- Peter M U Ung
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Masahiro Sonoshita
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Alex P Scopton
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Arvin C Dar
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Ross L Cagan
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
10
|
Wu Q, Kumar N, Velagala V, Zartman JJ. Tools to reverse-engineer multicellular systems: case studies using the fruit fly. J Biol Eng 2019; 13:33. [PMID: 31049075 PMCID: PMC6480878 DOI: 10.1186/s13036-019-0161-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/07/2019] [Indexed: 01/08/2023] Open
Abstract
Reverse-engineering how complex multicellular systems develop and function is a grand challenge for systems bioengineers. This challenge has motivated the creation of a suite of bioengineering tools to develop increasingly quantitative descriptions of multicellular systems. Here, we survey a selection of these tools including microfluidic devices, imaging and computer vision techniques. We provide a selected overview of the emerging cross-talk between engineering methods and quantitative investigations within developmental biology. In particular, the review highlights selected recent examples from the Drosophila system, an excellent platform for understanding the interplay between genetics and biophysics. In sum, the integrative approaches that combine multiple advances in these fields are increasingly necessary to enable a deeper understanding of how to analyze both natural and synthetic multicellular systems.
Collapse
Affiliation(s)
- Qinfeng Wu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Vijay Velagala
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Jeremiah J. Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
11
|
Villegas SN. One hundred years of Drosophila cancer research: no longer in solitude. Dis Model Mech 2019; 12:12/4/dmm039032. [PMID: 30952627 PMCID: PMC6505481 DOI: 10.1242/dmm.039032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
When Mary Stark first described the presence of tumours in the fruit fly Drosophila melanogaster in 1918, would she ever have imagined that flies would become an invaluable organism for modelling and understanding oncogenesis? And if so, would she have expected it to take 100 years for this model to be fully accredited? This Special Article summarises the efforts and achievements of Drosophilists to establish the fly as a valid model in cancer research through different scientific periods.
Collapse
Affiliation(s)
- Santiago Nahuel Villegas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernandez (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| |
Collapse
|
12
|
Stefana MI, Driscoll PC, Obata F, Pengelly AR, Newell CL, MacRae JI, Gould AP. Developmental diet regulates Drosophila lifespan via lipid autotoxins. Nat Commun 2017; 8:1384. [PMID: 29123106 PMCID: PMC5680271 DOI: 10.1038/s41467-017-01740-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022] Open
Abstract
Early-life nourishment exerts long-term influences upon adult physiology and disease risk. These lasting effects of diet are well established but the underlying mechanisms are only partially understood. Here we show that restricting dietary yeast during Drosophila development can, depending upon the subsequent adult environment, more than double median lifespan. Developmental diet acts via a long-term influence upon the adult production of toxic molecules, which we term autotoxins, that are shed into the environment and shorten the lifespan of both sexes. Autotoxins are synthesised by oenocytes and some of them correspond to alkene hydrocarbons that also act as pheromones. This study identifies a mechanism by which the developmental dietary history of an animal regulates its own longevity and that of its conspecific neighbours. It also has important implications for the design of lifespan experiments as autotoxins can influence the regulation of longevity by other factors including diet, sex, insulin signalling and population density.
Collapse
Affiliation(s)
- M Irina Stefana
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Paul C Driscoll
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Fumiaki Obata
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | | | - Clare L Newell
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - James I MacRae
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Alex P Gould
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
13
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017; 6. [PMID: 28892296 DOI: 10.1002/adhm.201700258] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Approaches to increase the efficiency in developing drugs and diagnostics tools, including new drug delivery and diagnostic technologies, are needed for improved diagnosis and treatment of major diseases and health problems such as cancer, inflammatory diseases, chronic wounds, and antibiotic resistance. Development within several areas of research ranging from computational sciences, material sciences, bioengineering to biomedical sciences and bioimaging is needed to realize innovative drug development and diagnostic (DDD) approaches. Here, an overview of recent progresses within key areas that can provide customizable solutions to improve processes and the approaches taken within DDD is provided. Due to the broadness of the area, unfortunately all relevant aspects such as pharmacokinetics of bioactive molecules and delivery systems cannot be covered. Tailored approaches within (i) bioinformatics and computer-aided drug design, (ii) nanotechnology, (iii) novel materials and technologies for drug delivery and diagnostic systems, and (iv) disease models to predict safety and efficacy of medicines under development are focused on. Current developments and challenges ahead are discussed. The broad scope reflects the multidisciplinary nature of the field of DDD and aims to highlight the convergence of biological, pharmaceutical, and medical disciplines needed to meet the societal challenges of the 21st century.
Collapse
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
14
|
Sahlgren C, Meinander A, Zhang H, Cheng F, Preis M, Xu C, Salminen TA, Toivola D, Abankwa D, Rosling A, Karaman DŞ, Salo-Ahen OMH, Österbacka R, Eriksson JE, Willför S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N. Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems. Adv Healthc Mater 2017. [DOI: 10.1002/adhm.201700258 10.1002/adhm.201700258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Affiliation(s)
- Cecilia Sahlgren
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Annika Meinander
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Hongbo Zhang
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Fang Cheng
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
| | - Maren Preis
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Chunlin Xu
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Tiina A. Salminen
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Diana Toivola
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Center for Disease Modeling; University of Turku; FI-20520 Turku Finland
| | - Daniel Abankwa
- Department of Biomedical Engineering; Technical University of Eindhoven; 5613 DR Eindhoven Netherlands
| | - Ari Rosling
- Faculty of Science and Engineering; Polymer Technologies; Åbo Akademi University; FI-20500 Turku Finland
| | - Didem Şen Karaman
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Outi M. H. Salo-Ahen
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Ronald Österbacka
- Faculty of Science and Engineering; Physics; Åbo Akademi University; FI-20500 Turku Finland
| | - John E. Eriksson
- Faculty of Science and Engineering; Cell Biology; Åbo Akademi University; FI-20520 Turku Finland
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; FI-20520 Turku Finland
| | - Stefan Willför
- Faculty of Science and Engineering; Natural Materials Technology; Åbo Akademi University; FI-20500 Turku Finland
| | - Ion Petre
- Faculty of Science and Engineering; Computer Science; Åbo Akademi University; FI-20500 Turku Finland
| | - Jouko Peltonen
- Faculty of Science and Engineering; Physical Chemistry; Åbo Akademi University; FI-20500 Turku Finland
| | - Reko Leino
- Faculty of Science and Engineering; Organic Chemistry; Johan Gadolin Process Chemistry Centre; Åbo Akademi University; FI-20500 Turku Finland
| | - Mark Johnson
- Faculty of Science and Engineering; Structural Bioinformatics Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Jessica Rosenholm
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| | - Niklas Sandler
- Faculty of Science and Engineering; Pharmaceutical Sciences Laboratory; Åbo Akademi University; FI-20520 Turku Finland
| |
Collapse
|
15
|
Panchal K, Tiwari AK. Drosophila melanogaster "a potential model organism" for identification of pharmacological properties of plants/plant-derived components. Biomed Pharmacother 2017; 89:1331-1345. [PMID: 28320100 DOI: 10.1016/j.biopha.2017.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/09/2017] [Accepted: 03/01/2017] [Indexed: 12/18/2022] Open
Abstract
Plants/plant-derived components have been used from ancient times to treat/cure several human diseases. Plants and their parts possess several chemical components that play the vital role in the improvement of human health and their life expectancy. Allopathic medicines have been playing a key role in the treatment of several diseases. Though allopathic medicines provide fast relief, long time consumption cause serious health concerns such as hyperallergic reactions, liver damage, etc. So, the study of medicinal plants which rarely cause any side effect is very important to mankind. Plants contain many health benefit properties like antioxidant, anti-aging, neuroprotective, anti-genotoxic, anti-mutagenic and bioinsecticidal activity. Thus, identification of pharmacological properties of plants/plant-derived components are of utmost importance to be explored. Several model organisms have been used to identify the pharmacological properties of the different plants or active components therein and Drosophila is one of them. Drosophila melanogaster "fruit fly" is a well understood, high-throughput model organism being used more than 110 years to study the different biological aspects related to the development and diseases. Most of the developmental and cell signaling pathways and ∼75% human disease-related genes are conserved between human and Drosophila. Using Drosophila, one can easily analyze the pharmacological properties of plants/plant-derived components by performing several assays available with flies such as survivorship, locomotor, antioxidant, cell death, etc. The current review focuses on the potential of Drosophila melanogaster for the identification of medicinal/pharmacological properties associated with plants/plant-derived components.
Collapse
Affiliation(s)
- Komal Panchal
- Genetics & Developmental Biology Laboratory, School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research/IAR, Koba Institutional Area, Gandhinagar 382 007, Gujarat, India.
| | - Anand K Tiwari
- Genetics & Developmental Biology Laboratory, School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research/IAR, Koba Institutional Area, Gandhinagar 382 007, Gujarat, India.
| |
Collapse
|
16
|
Insulin and TOR signal in parallel through FOXO and S6K to promote epithelial wound healing. Nat Commun 2016; 7:12972. [PMID: 27713427 PMCID: PMC5059774 DOI: 10.1038/ncomms12972] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/22/2016] [Indexed: 12/18/2022] Open
Abstract
The TOR and Insulin/IGF signalling (IIS) network controls growth, metabolism and ageing. Although reducing TOR or insulin signalling can be beneficial for ageing, it can be detrimental for wound healing, but the reasons for this difference are unknown. Here we show that IIS is activated in the cells surrounding an epidermal wound in Drosophila melanogaster larvae, resulting in PI3K activation and redistribution of the transcription factor FOXO. Insulin and TOR signalling are independently necessary for normal wound healing, with FOXO and S6K as their respective effectors. IIS is specifically required in cells surrounding the wound, and the effect is independent of glycogen metabolism. Insulin signalling is needed for the efficient assembly of an actomyosin cable around the wound, and constitutively active myosin II regulatory light chain suppresses the effects of reduced IIS. These findings may have implications for the role of insulin signalling and FOXO activation in diabetic wound healing.
Collapse
|
17
|
Caicedo JC, Singh S, Carpenter AE. Applications in image-based profiling of perturbations. Curr Opin Biotechnol 2016; 39:134-142. [PMID: 27089218 DOI: 10.1016/j.copbio.2016.04.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 12/19/2022]
Abstract
A dramatic shift has occurred in how biologists use microscopy images. Whether experiments are small-scale or high-throughput, automatically quantifying biological properties in images is now widespread. We see yet another revolution under way: a transition towards using automated image analysis to not only identify phenotypes a biologist specifically seeks to measure ('screening') but also as an unbiased and sensitive tool to capture a wide variety of subtle features of cell (or organism) state ('profiling'). Mapping similarities among samples using image-based (morphological) profiling has tremendous potential to transform drug discovery, functional genomics, and basic biological research. Applications include target identification, lead hopping, library enrichment, functionally annotating genes/alleles, and identifying small molecule modulators of gene activity and disease-specific phenotypes.
Collapse
Affiliation(s)
- Juan C Caicedo
- Imaging Platform of the Broad Institute of Harvard and Massachusetts Institute of Technology, 415 Main Street, Cambridge, MA, USA; Fundación Universitaria Konrad Lorenz, Bogotá, Colombia
| | - Shantanu Singh
- Imaging Platform of the Broad Institute of Harvard and Massachusetts Institute of Technology, 415 Main Street, Cambridge, MA, USA
| | - Anne E Carpenter
- Imaging Platform of the Broad Institute of Harvard and Massachusetts Institute of Technology, 415 Main Street, Cambridge, MA, USA.
| |
Collapse
|
18
|
|
19
|
Mohr SE. RNAi screening in Drosophila cells and in vivo. Methods 2014; 68:82-8. [PMID: 24576618 DOI: 10.1016/j.ymeth.2014.02.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 12/31/2022] Open
Abstract
Here, I discuss how RNAi screening can be used effectively to uncover gene function. Specifically, I discuss the types of high-throughput assays that can be done in Drosophila cells and in vivo, RNAi reagent design and available reagent collections, automated screen pipelines, analysis of screen results, and approaches to RNAi results verification.
Collapse
Affiliation(s)
- Stephanie E Mohr
- Drosophila RNAi Screening Center, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States.
| |
Collapse
|
20
|
Liu B, Campo EM, Bossing T. Drosophila embryos as model to assess cellular and developmental toxicity of multi-walled carbon nanotubes (MWCNT) in living organisms. PLoS One 2014; 9:e88681. [PMID: 24558411 PMCID: PMC3928276 DOI: 10.1371/journal.pone.0088681] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 01/09/2014] [Indexed: 02/07/2023] Open
Abstract
Different toxicity tests for carbon nanotubes (CNT) have been developed to assess their impact on human health and on aquatic and terrestrial animal and plant life. We present a new model, the fruit fly Drosophila embryo offering the opportunity for rapid, inexpensive and detailed analysis of CNTs toxicity during embryonic development. We show that injected DiI labelled multi-walled carbon nanotubes (MWCNTs) become incorporated into cells in early Drosophila embryos, allowing the study of the consequences of cellular uptake of CNTs on cell communication, tissue and organ formation in living embryos. Fluorescently labelled subcellular structures showed that MWCNTs remained cytoplasmic and were excluded from the nucleus. Analysis of developing ectodermal and neural stem cells in MWCNTs injected embryos revealed normal division patterns and differentiation capacity. However, an increase in cell death of ectodermal but not of neural stem cells was observed, indicating stem cell-specific vulnerability to MWCNT exposure. The ease of CNT embryo injections, the possibility of detailed morphological and genomic analysis and the low costs make Drosophila embryos a system of choice to assess potential developmental and cellular effects of CNTs and test their use in future CNT based new therapies including drug delivery.
Collapse
Affiliation(s)
- Boyin Liu
- School of Biological Sciences, University of Bangor, Bangor, United Kingdom
| | - Eva M. Campo
- School of Electronic Engineering, University of Bangor, Bangor, United Kingdom
| | - Torsten Bossing
- School of Biological Sciences, University of Bangor, Bangor, United Kingdom
| |
Collapse
|
21
|
Demir E, Turna F, Vales G, Kaya B, Creus A, Marcos R. In vivo genotoxicity assessment of titanium, zirconium and aluminium nanoparticles, and their microparticulated forms, in Drosophila. CHEMOSPHERE 2013; 93:2304-2310. [PMID: 24095613 DOI: 10.1016/j.chemosphere.2013.08.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 07/18/2013] [Accepted: 08/10/2013] [Indexed: 06/02/2023]
Abstract
As in vivo system, we propose Drosophila melanogaster as a useful model for study the genotoxic risks associated with nanoparticle exposure. In this study we have carried out a genotoxic evaluation of titanium dioxide (TiO2), zirconium oxide (ZrO2) and aluminium oxide (Al2O3) nanoparticles and their microparticulated forms in D. melanogaster by using the wing somatic mutation and recombination assay. This assay is based on the principle that loss of heterozygosis and the corresponding expression of the suitable recessive markers, multiple wing hairs and flare-3, can lead to the formation of mutant clones in treated larvae, which are expressed as mutant spots on the wings of adult flies. Third instar larvae were feed with TiO2, ZrO2 and Al2O3 nanoparticles, and their microparticulated forms, at concentrations ranging from 0.1 to 10mM. Although a certain level of aggregation/agglomeration was observed in solution, it must be noted than the constant digging activity of larvae ensures that treated medium pass constantly through the digestive tract ensuring exposure. The results showed that no significant increases in the frequency of all spots (e.g. small single, large single, twin, total mwh and total spots) were observed, indicating that these nanoparticles were not able to induce genotoxic activity in the wing spot assay of D. melanogaster. Negative data were also obtained with the microparticulated forms. This indicates that the nanoparticulated form of the selected nanomaterials does not modify the potential genotoxicity of their microparticulated versions. These in vivo results contribute to increase the genotoxicity database on the TiO2, ZrO2 and Al2O3 nanoparticles.
Collapse
Affiliation(s)
- Eşref Demir
- Akdeniz University, Faculty of Sciences, Department of Biology, 07058 Antalya, Turkey
| | | | | | | | | | | |
Collapse
|
22
|
Neckameyer WS, Argue KJ. Comparative approaches to the study of physiology: Drosophila as a physiological tool. Am J Physiol Regul Integr Comp Physiol 2012; 304:R177-88. [PMID: 23220476 DOI: 10.1152/ajpregu.00084.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Numerous studies have detailed the extensive conservation of developmental signaling pathways between the model system, Drosophila melanogaster, and mammalian models, but researchers have also profited from the unique and highly tractable genetic tools available in this system to address critical questions in physiology. In this review, we have described contributions that Drosophila researchers have made to mathematical dynamics of pattern formation, cardiac pathologies, the way in which pain circuits are integrated to elicit responses from sensation, as well as the ways in which gene expression can modulate diverse behaviors and shed light on human cognitive disorders. The broad and diverse array of contributions from Drosophila underscore its translational relevance to modeling human disease.
Collapse
Affiliation(s)
- Wendi S Neckameyer
- Dept. of Pharmacological and Physiological Science, St. Louis Univ. School of Medicine, St. Louis, MO 63104, USA.
| | | |
Collapse
|